Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1-2 study.
Abstract
BACKGROUND:Internal tandem duplication mutations in FLT3 are common in acute myeloid
leukaemia and are associated with rapid relapse and short overall survival. The clinical
benefit of FLT3 inhibitors in patients with acute myeloid leukaemia has been limited
by rapid generation of resistance mutations, particularly in codon Asp835 (D835).
We aimed to assess the highly selective oral FLT3 inhibitor gilteritinib in patients
with relapsed or refractory acute myeloid leukaemia. METHODS:In this phase 1-2 trial,
we enrolled patients aged 18 years or older with acute myeloid leukaemia who either
were refractory to induction therapy or had relapsed after achieving remission with
previous treatment. Patients were enrolled into one of seven dose-escalation or dose-expansion
cohorts assigned to receive once-daily doses of oral gilteritinib (20 mg, 40 mg, 80
mg, 120 mg, 200 mg, 300 mg, or 450 mg). Cohort expansion was based on safety and tolerability,
FLT3 inhibition in correlative assays, and antileukaemic activity. Although the presence
of an FLT3 mutation was not an inclusion criterion, we required ten or more patients
with locally confirmed FLT3 mutations (FLT3mut+) to be enrolled in expansion cohorts
at each dose level. On the basis of emerging findings, we further expanded the 120
mg and 200 mg dose cohorts to include FLT3mut+ patients only. The primary endpoints
were the safety, tolerability, and pharmacokinetics of gilteritinib. Safety and tolerability
were assessed in the safety analysis set (all patients who received at least one dose
of gilteritinib). Responses were assessed in the full analysis set (all patients who
received at least one dose of study drug and who had at least one datapoint post-treatment).
Pharmacokinetics were assessed in a subset of the safety analysis set for which sufficient
data for concentrations of gilteritinib in plasma were available to enable derivation
of one or more pharmacokinetic variables. This study is registered with ClinicalTrials.gov,
number NCT02014558, and is ongoing. FINDINGS:Between Oct 15, 2013, and Aug 27, 2015,
252 adults with relapsed or refractory acute myeloid leukaemia received oral gilteritinib
once daily in one of seven dose-escalation (n=23) or dose-expansion (n=229) cohorts.
Gilteritinib was well tolerated; the maximum tolerated dose was established as 300
mg/day when two of three patients enrolled in the 450 mg dose-escalation cohort had
two dose-limiting toxicities (grade 3 diarrhoea and grade 3 elevated aspartate aminotransferase).
The most common grade 3-4 adverse events irrespective of relation to treatment were
febrile neutropenia (97 [39%] of 252), anaemia (61 [24%]), thrombocytopenia (33 [13%]),
sepsis (28 [11%]), and pneumonia (27 [11%]). Commonly reported treatment-related adverse
events were diarrhoea (92 [37%] of 252]), anaemia (86 [34%]), fatigue (83 [33%]),
elevated aspartate aminotransferase (65 [26%]), and increased alanine aminotransferase
(47 [19%]). Serious adverse events occurring in 5% or more of patients were febrile
neutropenia (98 [39%] of 252; five related to treatment), progressive disease (43
[17%]), sepsis (36 [14%]; two related to treatment), pneumonia (27 [11%]), acute renal
failure (25 [10%]; five related to treatment), pyrexia (21 [8%]; three related to
treatment), bacteraemia (14 [6%]; one related to treatment), and respiratory failure
(14 [6%]). 95 people died in the safety analysis set, of which seven deaths were judged
possibly or probably related to treatment (pulmonary embolism [200 mg/day], respiratory
failure [120 mg/day], haemoptysis [80 mg/day], intracranial haemorrhage [20 mg/day],
ventricular fibrillation [120 mg/day], septic shock [80 mg/day], and neutropenia [120
mg/day]). An exposure-related increase in inhibition of FLT3 phosphorylation was noted
with increasing concentrations in plasma of gilteritinib. In-vivo inhibition of FLT3
phosphorylation occurred at all dose levels. At least 90% of FLT3 phosphorylation
inhibition was seen by day 8 in most patients receiving a daily dose of 80 mg or higher.
100 (40%) of 249 patients in the full analysis set achieved a response, with 19 (8%)
achieving complete remission, ten (4%) complete remission with incomplete platelet
recovery, 46 (18%) complete remission with incomplete haematological recovery, and
25 (10%) partial remission INTERPRETATION: Gilteritinib had a favourable safety profile
and showed consistent FLT3 inhibition in patients with relapsed or refractory acute
myeloid leukaemia. These findings confirm that FLT3 is a high-value target for treatment
of relapsed or refractory acute myeloid leukaemia; based on activity data, gilteritinib
at 120 mg/day is being tested in phase 3 trials. FUNDING:Astellas Pharma, National
Cancer Institute (Leukemia Specialized Program of Research Excellence grant), Associazione
Italiana Ricerca sul Cancro.
Type
Journal articleSubject
Blood PlateletsHumans
Recurrence
Aniline Compounds
Pyrazines
Antineoplastic Agents
Retreatment
Maximum Tolerated Dose
Phosphorylation
Aged
Middle Aged
Female
Male
fms-Like Tyrosine Kinase 3
Leukemia, Myeloid, Acute
Permalink
https://hdl.handle.net/10161/19547Published Version (Please cite this version)
10.1016/S1470-2045(17)30416-3Publication Info
Perl, Alexander E; Altman, Jessica K; Cortes, Jorge; Smith, Catherine; Litzow, Mark;
Baer, Maria R; ... Levis, Mark (2017). Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid
leukaemia: a multicentre, first-in-human, open-label, phase 1-2 study. The Lancet. Oncology, 18(8). pp. 1061-1075. 10.1016/S1470-2045(17)30416-3. Retrieved from https://hdl.handle.net/10161/19547.This is constructed from limited available data and may be imprecise. To cite this
article, please review & use the official citation provided by the journal.
Collections
More Info
Show full item recordScholars@Duke
Harry Paul Erba
Instructor in the Department of Medicine
I am a clinical investigator in the Division of Hematologic Malignancies and Cellular
Therapy in the Department of Medicine. I serve as Director of the Leukemia Program
and Director of Phase I Development in Hematologic Malignancies. I am also the Chair
of the SWOG Leukemia Committee. I am interested in the clinical development of novel
therapies for acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative
neoplasms (such as chronic myeloid leukemia, polycythemia v

Articles written by Duke faculty are made available through the campus open access policy. For more information see: Duke Open Access Policy
Rights for Collection: Scholarly Articles
Works are deposited here by their authors, and represent their research and opinions, not that of Duke University. Some materials and descriptions may include offensive content. More info